Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
Nat Immunol ; 21(7): 746-755, 2020 07.
Article in English | MEDLINE | ID: mdl-32514064

ABSTRACT

Plasma membranes of animal cells are enriched for cholesterol. Cholesterol-dependent cytolysins (CDCs) are pore-forming toxins secreted by bacteria that target membrane cholesterol for their effector function. Phagocytes are essential for clearance of CDC-producing bacteria; however, the mechanisms by which these cells evade the deleterious effects of CDCs are largely unknown. Here, we report that interferon (IFN) signals convey resistance to CDC-induced pores on macrophages and neutrophils. We traced IFN-mediated resistance to CDCs to the rapid modulation of a specific pool of cholesterol in the plasma membrane of macrophages without changes to total cholesterol levels. Resistance to CDC-induced pore formation requires the production of the oxysterol 25-hydroxycholesterol (25HC), inhibition of cholesterol synthesis and redistribution of cholesterol to an esterified cholesterol pool. Accordingly, blocking the ability of IFN to reprogram cholesterol metabolism abrogates cellular protection and renders mice more susceptible to CDC-induced tissue damage. These studies illuminate targeted regulation of membrane cholesterol content as a host defense strategy.


Subject(s)
Bacterial Infections/immunology , Bacterial Toxins/immunology , Hydroxycholesterols/metabolism , Interferons/isolation & purification , Phagocytes/immunology , Streptolysins/immunology , Animals , Bacteria/immunology , Bacteria/metabolism , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cell Membrane/metabolism , Cell Membrane Permeability/immunology , Cells, Cultured , Disease Models, Animal , Disease Susceptibility/immunology , Female , Host Microbial Interactions/immunology , Humans , Intravital Microscopy , Male , Mice , Mice, Transgenic , Phagocytes/cytology , Phagocytes/metabolism , Primary Cell Culture , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Streptolysins/administration & dosage , Streptolysins/metabolism
2.
Biochem Biophys Res Commun ; 495(1): 1432-1439, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29158087

ABSTRACT

Functional abnormal airway epithelial cells, along with activated inflammatory cells, resulting in chronic airway inflammation, are considered as the characteristic of asthma. Fatty Acid Binding Protein 4 (FABP4) takes part in glucose and lipid homeostasis, and also have an important role in allergic airway inflammation. However, whether FABP4 influence barrier function of airway epithelial cells is unknown. In vivo, a HDM-induced murine model of asthma was obtained to assessed airway inflammation and protein expression of E-cadherin and Forkhead Box M1 (FoxM1). In vitro, 16-HBE was cultured and was treated with hrFABP4, siFABP4, FABPF4 inhibitor BMS, or FoxM1 inhibitor RCM-1. IL-4, IL-5, and IL-13 level was determined by ELISA. Transepithelial electrical resistance (TER), paracellular permeability and E-cadherin-special immunofluorescence were measured to value airway epithelial barrier function. Intracellular ROS production was determined by DCF-DA fluorescence. FABP4 inhibitor BMS alleviate airway inflammation and destruction of E-cad in allergic mouse. Treatment with HDM or hrFABP4 aggravated inflammatory response, damaged airway epithelial barrier, which could be inhibited by siFABP4 and BMS. Treatment with HDM or hrFABP4 also enhanced levels of FoxM1, and Inhibited FoxM1 suppressed HDM- and hrFABP4-induced inflammation and airway epithelial barrier dysfunction. In addition, H2O2 promoted FoxM1 expression, HDM and hrFABP4 induced-FoxM1 could be inhibited by NAC, leading to decreased inflammation and improved airway epithelial barrier. Upregulated ROS induced by FABP4 was of significance in activating FoxM1 leading to airway inflammation and epithelial barrier dysfunction.


Subject(s)
Alveolar Epithelial Cells/immunology , Asthma/immunology , Cell Membrane Permeability/immunology , Fatty Acid-Binding Proteins/immunology , Forkhead Box Protein M1/immunology , Reactive Oxygen Species/immunology , Respiratory Mucosa/immunology , Animals , Asthma/pathology , Male , Mice , Mice, Inbred BALB C , Respiratory Mucosa/pathology
3.
Brain Behav Immun ; 65: 68-89, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28676349

ABSTRACT

When related to central nervous system (CNS) health and disease, brain mast cells (MCs) can be a source of either beneficial or deleterious signals acting on neural cells. We review the current state of knowledge about molecular interactions between MCs and glia in neurodegenerative diseases such as Multiple Sclerosis, Alzheimer's disease, Amyotrophic Lateral Sclerosis, Parkinson's disease, Epilepsy. We also discuss the influence on MC actions evoked by the host microbiota, which has a profound effect on the host immune system, inducing important consequences in neurodegenerative disorders. Gut dysbiosis, reduced intestinal motility and increased intestinal permeability, that allow bacterial products to circulate and pass through the blood-brain barrier, are associated with neurodegenerative disease. There are differences between the microbiota of neurologic patients and healthy controls. Distinguishing between cause and effect is a challenging task, and the molecular mechanisms whereby remote gut microbiota can alter the brain have not been fully elucidated. Nevertheless, modulation of the microbiota and MC activation have been shown to promote neuroprotection. We review this new information contributing to a greater understanding of MC-microbiota-neural cells interactions modulating the brain, behavior and neurodegenerative processes.


Subject(s)
Mast Cells/immunology , Mast Cells/physiology , Neurodegenerative Diseases/immunology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/physiology , Brain/immunology , Cell Communication , Cell Membrane Permeability/immunology , Central Nervous System/immunology , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Gastrointestinal Motility/immunology , Humans , Intestines/microbiology , Mast Cells/microbiology , Microbiota , Neurodegenerative Diseases/microbiology , Neuroglia/immunology
4.
Respir Res ; 17: 2, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26739349

ABSTRACT

BACKGROUND: COPD patients have a higher risk of pneumonia when treated with fluticasone propionate (FP) than with placebo, and a lower risk with budesonide (BUD). We hypothesized that BUD and FP differentially affect the mucosal barrier in response to viral infection and/or cigarette smoke. METHODS: We assessed protective effects of equivalent concentrations of BUD and FP on cytokine production and barrier function (electrical resistance) in human bronchial epithelial 16HBE cells and primary bronchial epithelial cells (PBECs) upon exposure to viral mimetic poly-(I:C) and/or cigarette smoke extract (CSE) or epidermal growth factor (EGF). RESULTS: BUD and FP were equally effective in suppressing poly-(I:C)- and/or CSE-induced IL-8 secretion in 16HBE and PBECs. Poly-(I:C) substantially decreased electrical resistance in 16HBE cells and both BUD and FP fully counteracted this effect. However, FP hardly affected 16HBE barrier dysfunction induced by CSE with/without poly-(I:C), whereas BUD (16 nM) provided full protection, an effect likely mediated by affecting EGFR-downstream target GSK-3ß. Similarly, BUD, but not FP, significantly improved CSE-induced barrier dysfunction in PBECs. Finally, BUD, but not FP, exerted a modest but significant protective effect against Streptococcus Pneumoniae-induced barrier dysfunction, and BUD, but not FP, prevented cellular adhesion and/or internalization of these bacteria induced by poly-(I:C) in 16HBE. CONCLUSIONS: Collectively, both BUD and FP efficiently control epithelial pro-inflammatory responses and barrier function upon mimicry of viral infection. Of potential clinical relevance, BUD more effectively counteracted CSE-induced barrier dysfunction, reinforcing the epithelial barrier and potentially limiting access of pathogens upon smoking in vivo.


Subject(s)
Bronchi/immunology , Budesonide/administration & dosage , Epithelial Cells/immunology , Epithelial Cells/virology , Fluticasone/administration & dosage , Poly C/immunology , Bronchi/drug effects , Bronchi/virology , Bronchodilator Agents/administration & dosage , Cell Line , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/immunology , Cytokines/immunology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Humans , Rhinovirus/drug effects , Rhinovirus/physiology , Tars
5.
Circ Res ; 114(4): 637-49, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24366170

ABSTRACT

RATIONALE: Natural killer (NK) cells are lymphocytes of the innate immune system that play specialized and niche-specific roles in distinct organs. OBJECTIVE: We investigated the possible function of NK cells in the pathogenesis of congestive heart failure after myocardial infarction. METHODS AND RESULTS: Depletion of NK cells from mice had little effect on cytokine expression (tumor necrosis factor-α, interleukin [IL]-6, and IL-1ß), neutrophil and macrophage infiltration into infarcted myocardium, or left ventricular remodeling after myocardial infarction. However, these mice exhibited severe respiratory distress associated with protein-rich, high-permeability alveolar edema accompanied by neutrophil infiltration. In addition, there were 20-fold more NK cells in the mouse lungs than in heart, and these cells were accumulated around the vasculature. CD107a-positive and interferon-γ-positive cell populations were unchanged, whereas IL-10-positive populations increased. Adoptive transfer of NK cells from wild-type mice, but not from IL-10 knockout mice, into the NK cell-depleted mice rescued the respiratory phenotype. IL-1ß-mediated dextran leakage from a lung endothelial cell monolayer was also blocked by coculture with NK cells from wild-type mice but not from IL-10 knockout mice. CONCLUSIONS: This study is the first to identify a critical role for lung NK cells in protecting lung from the development of cardiogenic pulmonary edema after myocardial infarction.


Subject(s)
Endothelial Cells/immunology , Killer Cells, Natural/immunology , Myocardial Infarction/immunology , Pneumonia/immunology , Pulmonary Alveoli/immunology , Pulmonary Edema/immunology , Adoptive Transfer , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Membrane Permeability/immunology , Female , Green Fluorescent Proteins/genetics , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Killer Cells, Natural/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/complications , Neutrophils/immunology , Neutrophils/pathology , Pneumonia/complications , Pneumonia/pathology , Pulmonary Alveoli/pathology , Pulmonary Edema/complications , Pulmonary Edema/pathology
6.
Xenotransplantation ; 22(4): 295-301, 2015.
Article in English | MEDLINE | ID: mdl-26031609

ABSTRACT

Endothelial cell activation and injury by the terminal pathway of complement is important in various pathobiological processes, including xenograft rejection. Protection against injury by human complement can be induced in porcine endothelial cells (ECs) with IL-4 and IL-13 through metabolic activation. However, despite this resistance, the complement-treated ECs were found to lose membrane permeability control assessed with the small molecule calcein. Therefore, to define the apparent discrepancy of permeability changes vis-à-vis the protection from killing, we now investigated whether IL-4 and IL-13 influence the release of the large cytoplasmic protein lactate dehydrogenase (LDH) in ECs incubated with complement or the pore-forming protein melittin. Primary cultures of ECs were pre-treated with IL-4 or IL-13 and then incubated with human serum as source of antibody and complement or melittin. Cell death was assessed using neutral red. Membrane permeability was quantitated measuring LDH release. We found that IL-4-/IL-13-induced protection of ECs from killing by complement or melittin despite loss of LDH in amounts similar to control ECs. However, the cytokine-treated ECs that were protected from killing rapidly regained effective control of membrane permeability. Moreover, the viability of the protected ECs was maintained for at least 2 days. We conclude that the protection induced by IL-4/IL-13 in ECs against lethal attack by complement or melittin is effective and durable despite severe initial impairment of membrane permeability. The metabolic changes responsible for protection allow the cells to repair the membrane injury caused by complement or melittin.


Subject(s)
Complement System Proteins/toxicity , Endothelial Cells/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Interleukin-13/administration & dosage , Interleukin-4/administration & dosage , Melitten/toxicity , Animals , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/immunology , Cytoplasm/metabolism , Cytotoxicity, Immunologic , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Swine , Transplantation, Heterologous/adverse effects , Transplantation, Heterologous/methods
7.
Gastroenterology ; 144(5): 1116-28, 2013 May.
Article in English | MEDLINE | ID: mdl-23391819

ABSTRACT

BACKGROUND & AIMS: IgG4-related cholangitis is a chronic inflammatory biliary disease that involves different parts of the pancreatobiliary system, but little is known about its mechanisms of pathogenesis. A T-helper (Th) 2 cell cytokine profile predominates in liver tissues from these patients. We investigated whether Th2 cytokines disrupt the barrier function of biliary epithelial cells (BECs) in patients with IgG4-related cholangitis. METHODS: We assessed the Th2 cytokine profile in bile samples and brush cytology samples from 16 patients with IgG4-related cholangitis and respective controls, and evaluated transcription of tight junction (TJ)-associated proteins in primary BECs from these patients. The effect of Th2 cytokines on TJ-mediated BEC barrier function and wound closure was examined by immunoblot, transepithelial resistance, charge-selective Na(+)/Cl(-) permeability, and 4-kDa dextran flux analyses. RESULTS: Bile samples from patients with IgG4-related cholangitis had significant increases in levels of Th2 cytokines, interleukin (IL)-4, and IL-5. IL-13 was not detected in bile samples, but polymerase chain reaction analysis of whole-brush cytology samples from patients with IgG4-related cholangitis revealed increased levels of IL-13 mRNA, compared with controls. BECs isolated from the brush cytology samples revealed decreased levels of claudin-1 and increased levels of claudin-2 mRNAs. In vitro, IL-4 and IL-13 significantly reduced TJ-associated BEC barrier function by activating claudin-2-mediated paracellular pore pathways. Th2 cytokines also impaired wound closure in BEC monolayers. CONCLUSIONS: Th2 cytokines predominate in bile samples from patients with IgG4-related cholangitis and disrupt the TJ-mediated BEC barrier in vitro. Subsequent increases in biliary leaks might contribute to the pathogenesis of chronic biliary inflammation in these patients.


Subject(s)
Bile/metabolism , Cell Membrane Permeability/immunology , Cholangitis/immunology , Cytokines/metabolism , Epithelial Cells/metabolism , Immunoglobulin G/immunology , Th2 Cells/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/metabolism , Blotting, Western , Cells, Cultured , Cholangitis/metabolism , Cholangitis/pathology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/pathology , Female , Humans , Immunity, Cellular , Male , Middle Aged , Tight Junctions
8.
PLoS Pathog ; 8(3): e1002584, 2012.
Article in English | MEDLINE | ID: mdl-22457619

ABSTRACT

Dengue virus causes ∼50-100 million infections per year and thus is considered one of the most aggressive arthropod-borne human pathogen worldwide. During its replication, dengue virus induces dramatic alterations in the intracellular membranes of infected cells. This phenomenon is observed both in human and vector-derived cells. Using high-resolution mass spectrometry of mosquito cells, we show that this membrane remodeling is directly linked to a unique lipid repertoire induced by dengue virus infection. Specifically, 15% of the metabolites detected were significantly different between DENV infected and uninfected cells while 85% of the metabolites detected were significantly different in isolated replication complex membranes. Furthermore, we demonstrate that intracellular lipid redistribution induced by the inhibition of fatty acid synthase, the rate-limiting enzyme in lipid biosynthesis, is sufficient for cell survival but is inhibitory to dengue virus replication. Lipids that have the capacity to destabilize and change the curvature of membranes as well as lipids that change the permeability of membranes are enriched in dengue virus infected cells. Several sphingolipids and other bioactive signaling molecules that are involved in controlling membrane fusion, fission, and trafficking as well as molecules that influence cytoskeletal reorganization are also up regulated during dengue infection. These observations shed light on the emerging role of lipids in shaping the membrane and protein environments during viral infections and suggest membrane-organizing principles that may influence virus-induced intracellular membrane architecture.


Subject(s)
Aedes/virology , Dengue Virus/physiology , Lipid Metabolism , Aedes/cytology , Animals , Cell Membrane Permeability/immunology , Cell Membrane Permeability/physiology , Cells, Cultured , Dengue Virus/immunology , Dengue Virus/pathogenicity , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/metabolism , Homeostasis , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Intracellular Membranes/immunology , Intracellular Membranes/virology , Mass Spectrometry , Principal Component Analysis , Virus Replication
9.
J Immunol ; 188(2): 541-7, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22147765

ABSTRACT

Histamine is a biogenic amine that mediates multiple physiological processes, including immunomodulatory effects in allergic and inflammatory reactions, and also plays a key regulatory role in experimental allergic encephalomyelitis, the autoimmune model of multiple sclerosis. The pleiotropic effects of histamine are mediated by four G protein-coupled receptors, as follows: Hrh1/H(1)R, Hrh2/H(2)R, Hrh3/H(3)R, and Hrh4/H(4)R. H(4)R expression is primarily restricted to hematopoietic cells, and its role in autoimmune inflammatory demyelinating disease of the CNS has not been studied. In this study, we show that, compared with wild-type mice, animals with a disrupted Hrh4 (H(4)RKO) develop more severe myelin oligodendrocyte glycoprotein (MOG)(35\x{2013}55)-induced experimental allergic encephalomyelitis. Mechanistically, we also show that H(4)R plays a role in determining the frequency of T regulatory (T(R)) cells in secondary lymphoid tissues, and regulates T(R) cell chemotaxis and suppressor activity. Moreover, the lack of H(4)R leads to an impairment of an anti-inflammatory response due to fewer T(R) cells in the CNS during the acute phase of the disease and an increase in the proportion of Th17 cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Receptors, G-Protein-Coupled/physiology , Receptors, Histamine/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , Blood-Brain Barrier/immunology , CD4 Lymphocyte Count , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Glycoproteins/administration & dosage , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Neurons/immunology , Neurons/pathology , Peptide Fragments/administration & dosage , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine/deficiency , Receptors, Histamine/genetics , Receptors, Histamine H4 , Severity of Illness Index , T-Lymphocytes, Regulatory/metabolism
10.
J Immunol ; 189(2): 629-37, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706076

ABSTRACT

The neutrophil formyl peptide receptors, FPR1 and FPR2, play critical roles for inflammatory reactions, and receptor-specific antagonists/inhibitors can possibly be used to facilitate the resolution of pathological inflammatory reactions. A 10-aa-long rhodamine-linked and membrane-permeable peptide inhibitor (PBP10) has such a potential. This FPR2 selective inhibitor adopts a phosphatidylinositol 4,5-bisphosphate-binding sequence in the cytoskeletal protein gelsolin. A core peptide, RhB-QRLFQV, is identified that displays inhibitory effects as potent as the full-length molecule. The phosphatidylinositol 4,5-bisphosphate-binding capacity of PBP10 was not in its own sufficient for inhibition. A receptor in which the presumed cytoplasmic signaling C-terminal tail of FPR2 was replaced with that of FPR1 retained the PBP10 sensitivity, suggesting that the tail of FPR2 was not on its own critical for inhibition. This gains support from the fact that the effect of cell-penetrating lipopeptide (a pepducin), suggested to act primarily through the third intracellular loop of FPR2, was significantly inhibited by PBP10. The third intracellular loops of FPR1 and FPR2 differ in only two amino acids, but an FPR2 mutant in which these two amino acids were replaced by those present in FPR1 retained the PBP10 sensitivity. In summary, we conclude that the inhibitory activity on neutrophil function of PBP10 is preserved in the core sequence RhB-QRLFQV and that neither the third intracellular loop of FPR2 nor the cytoplasmic tail of the receptor alone is responsible for the specific inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Gelsolin/chemistry , Gelsolin/physiology , Peptides/chemistry , Peptides/physiology , Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/chemistry , Receptors, Lipoxin/physiology , Amino Acid Sequence , Cell Membrane Permeability/immunology , Dose-Response Relationship, Immunologic , Gelsolin/metabolism , HL-60 Cells , Humans , Molecular Sequence Data , Neutrophil Activation/immunology , Peptides/metabolism , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism
11.
J Immunol ; 189(6): 3085-91, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22888139

ABSTRACT

Clostridium difficile is a Gram-positive obligate anaerobic pathogen that causes pseudomembranous colitis in antibiotic-treated individuals. Commensal bacteria are known to have a significant role in the intestinal accumulation of C. difficile after antibiotic treatment, but little is known about how they affect host immunity during C. difficile infection. In this article, we report that C. difficile infection results in translocation of commensals across the intestinal epithelial barrier that is critical for neutrophil recruitment through the induction of an IL-1ß-mediated positive-feedback loop. Mice lacking ASC, an essential mediator of IL-1ß and IL-18 processing and secretion, were highly susceptible to C. difficile infection. ASC(-/-) mice exhibited enhanced translocation of commensals to multiple organs after C. difficile infection. Notably, ASC(-/-) mice exhibited impaired CXCL1 production and neutrophil influx into intestinal tissues in response to C. difficile infection. The impairment in neutrophil recruitment resulted in reduced production of IL-1ß and CXCL1 but not IL-18. Importantly, translocated commensals were required for ASC/Nlrp3-dependent IL-1ß secretion by neutrophils. Mice lacking IL-1ß were deficient in inducing CXCL1 secretion, suggesting that IL-1ß is the dominant inducer of ASC-mediated CXCL1 production during C. difficile infection. These results indicate that translocated commensals play a crucial role in CXCL1-dependent recruitment of neutrophils to the intestine through an IL-1ß/NLRP3/ASC-mediated positive-feedback mechanism that is important for host survival and clearance of translocated commensals during C. difficile infection.


Subject(s)
Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/prevention & control , Interleukin-1beta/physiology , Symbiosis/immunology , Up-Regulation/immunology , Animals , Biological Transport, Active/genetics , Biological Transport, Active/immunology , Cell Communication/immunology , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Enterocolitis, Pseudomembranous/pathology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Survival Analysis , Up-Regulation/genetics
12.
Blood ; 117(17): 4519-29, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21378274

ABSTRACT

The anti-CD20 mAb rituximab has substantially improved the clinical outcome of patients with a wide range of B-cell malignancies. However, many patients relapse or fail to respond to rituximab, and thus there is intense investigation into the development of novel anti-CD20 mAbs with improved therapeutic efficacy. Although Fc-FcγR interactions appear to underlie much of the therapeutic success with rituximab, certain type II anti-CD20 mAbs efficiently induce programmed cell death (PCD), whereas rituximab-like type I anti-CD20 mAbs do not. Here, we show that the humanized, glycoengineered anti-CD20 mAb GA101 and derivatives harboring non-glycoengineered Fc regions are type II mAb that trigger nonapoptotic PCD in a range of B-lymphoma cell lines and primary B-cell malignancies. We demonstrate that GA101-induced cell death is dependent on actin reorganization, can be abrogated by inhibitors of actin polymerization, and is independent of BCL-2 overexpression and caspase activation. GA101-induced PCD is executed by lysosomes which disperse their contents into the cytoplasm and surrounding environment. Taken together, these findings reveal that GA101 is able to potently elicit actin-dependent, lysosomal cell death, which may potentially lead to improved clearance of B-cell malignancies in vivo.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Death/immunology , Lymphoma, B-Cell , Actins/drug effects , Actins/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived/pharmacology , Cathepsins/pharmacology , Cell Adhesion/immunology , Cell Line, Tumor , Cell Membrane Permeability/immunology , Drug Resistance, Neoplasm/immunology , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lysosomes/drug effects , Lysosomes/immunology , Rituximab
13.
Transfusion ; 53 Suppl 1: 80S-90S, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23301978

ABSTRACT

BACKGROUND: After major traumatic injury, patients often require multiple transfusions of fresh frozen plasma (FFP) to correct coagulopathy and to reduce bleeding. A spray-dried plasma (SDP) product has several logistical benefits over FFP use in trauma patients with coagulopathy. These benefits include ease of transport, stability at room temperature, and rapid reconstitution for infusion. Our past work suggests that FFP promotes endothelial stability by inhibiting endothelial permeability. STUDY DESIGN AND METHODS: The main goal of this project is to determine if solvent-detergent-treated SDP is equivalent to FFP in inhibiting vascular endothelial cell (EC) permeability and inflammation in vitro. Furthermore, this study aimed to determine if solvent-detergent treatment and spray drying of plasma alters the protective effects of FFP on EC function. The five groups tested in our studies are the following: 1) fresh frozen-thawed plasma (FFP); 2) solvent-detergent-treated FFP; 3) solvent-detergent-treated SDP; 4) lactated Ringer's solution; and 5) Hextend. RESULTS: This study demonstrates that in vitro SDP and FFP equivalently inhibit vascular EC permeability, EC adherens junction breakdown, and endothelial white blood cell binding, an effect that is independent of changes in Vascular Cell Adhesion Molecule 1, Intracellular Adhesion Molecule 1, or E-selectin expression on ECs. Solvent-detergent treatment of FFP does not alter the protective effects of FFP on endothelial cell function in vitro. CONCLUSION: These data suggest the equivalence of FFP and SDP on modulation of endothelial function and inflammation in vitro.


Subject(s)
Endothelial Cells/immunology , Plasma , Vasculitis/immunology , Vasculitis/therapy , Adherens Junctions/immunology , Cell Adhesion/immunology , Cell Membrane Permeability/immunology , E-Selectin/metabolism , Endothelial Cells/cytology , Freeze Drying , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Leukocytes/cytology , Leukocytes/immunology , Pulmonary Artery/cytology , Vascular Cell Adhesion Molecule-1/metabolism
14.
J Immunol ; 187(10): 5255-67, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21967897

ABSTRACT

Sepsis remains the leading cause of death in critically ill patients, despite modern advances in critical care. Intestinal barrier dysfunction may lead to secondary bacterial translocation and the development of the multiple organ dysfunction syndrome during sepsis. Cyclooxygenase (COX)-2 is highly upregulated in the intestine during sepsis, and we hypothesized that it may be critical in the maintenance of intestinal epithelial barrier function during peritonitis-induced polymicrobial sepsis. COX-2(-/-) and COX-2(+/+) BALB/c mice underwent cecal ligation and puncture (CLP) or sham surgery. Mice chimeric for COX-2 were derived by bone marrow transplantation and underwent CLP. C2BBe1 cells, an intestinal epithelial cell line, were treated with the COX-2 inhibitor NS-398, PGD(2), or vehicle and stimulated with cytokines. COX-2(-/-) mice developed exaggerated bacteremia and increased mortality compared with COX-2(+/+) mice following CLP. Mice chimeric for COX-2 exhibited the recipient phenotype, suggesting that epithelial COX-2 expression in the ileum attenuates bacteremia following CLP. Absence of COX-2 significantly increased epithelial permeability of the ileum and reduced expression of the tight junction proteins zonula occludens-1, occludin, and claudin-1 in the ileum following CLP. Furthermore, PGD(2) attenuated cytokine-induced hyperpermeability and zonula occludens-1 downregulation in NS-398-treated C2BBe1 cells. Our findings reveal that absence of COX-2 is associated with enhanced intestinal epithelial permeability and leads to exaggerated bacterial translocation and increased mortality during peritonitis-induced sepsis. Taken together, our results suggest that epithelial expression of COX-2 in the ileum is a critical modulator of tight junction protein expression and intestinal barrier function during sepsis.


Subject(s)
Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Sepsis/immunology , Sepsis/mortality , Animals , Bacteremia/enzymology , Bacteremia/immunology , Bacteremia/mortality , Caco-2 Cells , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Cyclooxygenase 2/biosynthesis , Female , Humans , Ileum/enzymology , Ileum/immunology , Ileum/microbiology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/enzymology , Peritonitis/immunology , Peritonitis/mortality , Sepsis/enzymology
15.
J Immunol ; 187(12): 6402-9, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22095714

ABSTRACT

In addition to its antibacterial activity, the cathelicidin-derived LL-37 peptide induces multiple immunomodulatory effects on host cells. Atomic force microscopy, F-actin staining with phalloidin, passage of FITC-conjugated dextran through a monolayer of lung epithelial cells, and assessment of bacterial outgrowth from cells subjected to Pseudomonas aeruginosa infection were used to determine LL-37's effect on epithelial cell mechanical properties, permeability, and bacteria uptake. A concentration-dependent increase in stiffness and F-actin content in the cortical region of A549 cells and primary human lung epithelial cells was observed after treatment with LL-37 (0.5-5 µM), sphingosine 1-phosphate (1 µM), or LPS (1 µg/ml) or infection with PAO1 bacteria. Other cationic peptides, such as RK-31, KR-20, or WLBU2, and the antibacterial cationic steroid CSA-13 did not reproduce the effect of LL-37. A549 cell pretreatment with WRW4, an antagonist of the transmembrane formyl peptide receptor-like 1 protein attenuated LL-37's ability to increase cell stiffness. The LL-37-mediated increase in cell stiffness was accompanied by a decrease in permeability and P. aeruginosa uptake by a confluent monolayer of polarized normal human bronchial epithelial cells. These results suggested that the antibacterial effect of LL-37 involves an LL-37-dependent increase in cell stiffness that prevents epithelial invasion by bacteria.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Cathelicidins/physiology , Cell Membrane Permeability/immunology , Cell Migration Inhibition/immunology , Lung/immunology , Pseudomonas aeruginosa/pathogenicity , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Amino Acid Sequence , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Migration Inhibition/drug effects , Cells, Cultured , Humans , Lung/cytology , Lung/drug effects , Molecular Sequence Data , Pseudomonas aeruginosa/drug effects , Respiratory Mucosa/drug effects
16.
J Immunol ; 186(6): 3770-8, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21317397

ABSTRACT

Dimeric IgA Abs contribute significantly to the humoral part of the mucosal immune system. However, their potential as immunotherapeutic agent has hardly been explored. In this article, we describe the production, purification, and functional evaluation of recombinant dimeric IgA against the epidermal growth factor receptor. Human joining chain-containing IgA was produced by nonadherent Chinese hamster ovarian (CHO)-K1 cells under serum-free conditions. Purification by anti-human κ and anti-His-tag affinity, as well as size exclusion chromatography, resulted in a homogenous preparation of highly pure IgA dimers. Functional studies demonstrated dimeric IgA to be at least as effective as monomeric IgA in triggering Ab-dependent cellular cytotoxicity by isolated monocytes or polymorphonuclear cell and in human whole-blood assays. Importantly, dimeric IgA was more effective in F(ab)-mediated killing mechanisms, such as inhibition of ligand binding, receptor downmodulation, and growth inhibition. Furthermore, only dimeric but not monomeric IgA or IgG was directionally transported by the polymeric Ig receptor through an epithelial cell monolayer. Together, these studies demonstrate that recombinant dimeric IgA Abs recruit a distinct repertoire of effector functions compared with monomeric IgA or IgG1 Abs.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/immunology , Immunoglobulin A/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/immunology , Cell Death/immunology , Cell Line , Cell Line, Tumor , Cell Membrane Permeability/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Cricetinae , Dogs , Humans , Immunoglobulin A/chemistry , Immunoglobulin A/metabolism , Immunoglobulin Isotypes/chemistry , Immunoglobulin Isotypes/pharmacology , Kidney/cytology , Kidney/immunology , Kidney/metabolism , Mice , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
17.
J Immunol ; 186(4): 2065-72, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21228349

ABSTRACT

To understand better the endogenous sources of MHC class I peptide ligands, we generated an antigenic reporter protein whose degradation is rapidly and reversibly controlled with Shield-1, a cell-permeant drug. Using this system, we demonstrate that defective ribosomal products (DRiPs) represent a major and highly efficient source of peptides and are completely resistant to our attempts to stabilize the protein. Although peptides also derive from nascent Shield-1-sensitive proteins and "retirees" created by Shield-1 withdrawal, these are much less efficient sources on a molar basis. We use this system to identify two drugs--each known to inhibit polyubiquitin chain disassembly--that selectively inhibit presentation of Shield-1-resistant DRiPs. These findings provide the initial evidence for distinct biochemical pathways for presentation of DRiPs versus retirees and implicate polyubiquitin chain disassembly or the actions of deubiquitylating enzymes as playing an important role in DRiP presentation.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Surveillance , Peptide Biosynthesis/immunology , Ribosomal Proteins/biosynthesis , Ribosomal Proteins/deficiency , Signal Transduction/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Female , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , H-2 Antigens/biosynthesis , H-2 Antigens/genetics , Immunologic Surveillance/drug effects , Immunologic Surveillance/genetics , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Ovalbumin/immunology , Ovalbumin/metabolism , Peptide Biosynthesis/drug effects , Peptide Biosynthesis/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Stability/drug effects , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Mar Drugs ; 11(9): 3197-208, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-24065161

ABSTRACT

Lipid A, the hydrophobic anchor of lipopolysaccharide (LPS), is an essential component in the outer membrane of Gram-negative bacteria. It can stimulate the innate immune system via Toll-like receptor 4/myeloid differentiation factor 2 (TLR4/MD2), leading to the release of inflammatory cytokines. In this study, six Escherichia coli strains which can produce lipid A with different acylation patterns were constructed; the influence of lipid A acylation pattern on the membrane permeability and innate immune stimulation has been systematically investigated. The lipid A species were isolated and identified by matrix assisted laser ionization desorption-time of flight/tandem mass spectrometry. N-Phenyl naphthylamine uptake assay and antibiotic susceptibility test showed that membrane permeability of these strains were different. The lower the number of acyl chains in lipid A, the stronger the membrane permeability. LPS purified from these strains were used to stimulate human or mouse macrophage cells, and different levels of cytokines were induced. Compared with wild type hexa-acylated LPS, penta-acylated, tetra-acylated and tri-acylated LPS induced lower levels of cytokines. These results suggest that the lipid A acylation pattern influences both the bacterial membrane permeability and innate immune stimulation. The results would be useful for redesigning the bacterial membrane structure and for developing lipid A vaccine adjuvant.


Subject(s)
Acylation/immunology , Cell Membrane Permeability/immunology , Escherichia coli/immunology , Escherichia coli/metabolism , Immunity, Innate/immunology , Lipid A/immunology , Lipid A/metabolism , Animals , Cell Membrane/immunology , Cell Membrane/metabolism , Cytokines/immunology , Cytokines/metabolism , Humans , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/metabolism , Mice
19.
J Exp Med ; 203(3): 541-52, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16505137

ABSTRACT

The initiating etiologic factor in Crohn's disease (CD) remains unclear. SAMP1/YitFc (SAMP) mice develop chronic ileitis similar to human CD. We used bone marrow chimeras to determine if SAMP ileitis results from a primary immunological defect or from dysregulated mucosal immunity secondary to intrinsic, nonhematopoietic (e.g., epithelial) dysfunction. SAMP mice receiving wild-type (AKR) BM developed severe ileitis, whereas SAMP BM did not confer ileitis to WT recipients. WT lymphocytes from reconstituted SAMP mice resembled native SAMP populations in regard to surface phenotype and cytokine production. Ilea from native SAMP mice and SAMP recipients of wild-type BM displayed decreased epithelial barrier resistance ex vivo and increased epithelial permeability in vivo compared to native WT mice and AKR recipients of SAMP BM. This permeability defect preceded the development of ileal inflammation, was present in the absence of commensal bacteria, and was accompanied by altered ileal mRNA expression of the tight junction proteins claudin-2 and occludin. Our results provide evidence that the primary defect conferring ileitis in SAMP mice originates from a nonhematopoietic source. Generation of pathogenic lymphocytes is a consequence of this defect and does not reflect intrinsic proinflammatory leukocyte properties. Decreased barrier function suggests that defects in the epithelium may represent the primary source of SAMP ileitis susceptibility.


Subject(s)
Crohn Disease/immunology , Gene Expression Regulation/immunology , Ileitis/immunology , Lymphocytes/immunology , Animals , Bacteria/immunology , Bone Marrow Transplantation , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Claudins , Crohn Disease/genetics , Crohn Disease/pathology , Cytokines/immunology , Disease Models, Animal , Epithelium/immunology , Epithelium/pathology , Gene Expression Regulation/genetics , Hematopoiesis/immunology , Humans , Ileitis/genetics , Ileitis/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lymphocytes/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Transgenic , Occludin
20.
J Immunol ; 185(5): 2968-79, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20656927

ABSTRACT

Peripheral blood monocytes represent the rapid response component of mononuclear phagocyte host defense, generating vigorous but finite antibacterial responses. We investigated the fate of highly purified primary human monocytes following phagocytosis of different bacteria. Exposure to high bacterial loads resulted in rapid loss of cell viability and decreased functional competence. Cell death typically involved classical apoptosis. Exposure to high numbers of Escherichia coli and Klebsiella pneumoniae induced nonapoptotic death with loss of cell membrane integrity, marked disruption of phagolysosomes, and caspase-1 activation, while a subset of cells also released caspase-1-regulated extracellular traps. Classical apoptosis increased if extracellular bacterial replication was reduced and decreased if intracellular ATP levels were reduced during these infections. Both classical apoptosis and the alternative forms of cell death allowed monocytes, whose functional competence was exhausted, to downregulate reactive oxygen species and proinflammatory cytokine responses. In contrast, sustained stimulation of glycolytic metabolism and mitochondrial oxidative phosphorylation, with associated hypoxia inducible factor-1alpha upregulation, maintained intracellular ATP levels and prolonged monocyte functional longevity, as assessed by maintenance of phagocytosis, reactive oxygen species production, and proinflammatory cytokine generation. Monocyte innate responses to bacteria are short-lived and are limited by an intrinsic program of apoptosis, a response that is subverted by overwhelming infection with E. coli and K. pneumoniae or bacterial stimulation of cell metabolism. In this regard, the fate of monocytes following bacterial challenge more closely resembles neutrophils than macrophages.


Subject(s)
Escherichia coli Infections/immunology , Immunity, Innate , Klebsiella Infections/immunology , Meningococcal Infections/immunology , Monocytes/immunology , Monocytes/pathology , Neisseriaceae Infections/immunology , Cell Death/immunology , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , DNA/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Humans , Klebsiella Infections/microbiology , Klebsiella Infections/pathology , Meningococcal Infections/microbiology , Meningococcal Infections/pathology , Monocytes/metabolism , Monocytes/microbiology , Neisseriaceae Infections/microbiology , Neisseriaceae Infections/pathology
SELECTION OF CITATIONS
SEARCH DETAIL